Abstract
Introduction
The mouse is an important, though imperfect, organism with which to model human disease and to discover and test novel drugs in a preclinical setting. Many experimental strategies have been used to discover new biological and molecular targets in the mouse, with the hopes of translating these discoveries into novel drugs to treat prostate cancer in humans. Modeling prostate cancer in the mouse, however, has been challenging, and often drugs that work in mice have failed in human trials.
Areas covered
Here we discuss similarities and differences between mice and men, types of mouse models that exist to model prostate cancer, practical questions one must ask when using a mouse as a model, and potential reasons that drugs do not often translate to humans. We will also discuss the current value in using mouse models for drug discovery to treat prostate cancer and what needs are still unmet in field.
Expert opinion
With proper planning and following practical guidelines by the researcher, the mouse is a powerful experimental tool. The field lacks genetically engineered metastatic models, and xenograft models do not allow for the study of the immune system during the metastatic process. There remain several important limitations to discovering and testing novel drugs in mice for eventual human use, but these can often be overcome. Overall, mouse modeling is an essential part of prostate cancer research and drug discovery. Emerging technologies and better and ever-increasing forms of communication are moving the field in a hopeful direction.
1. Introduction
Prostate cancer is the most diagnosed non-skin cancer in men in the United States, with approximately 230,000 new cases each year1. The 5-year survival rate for patients with localized prostate cancer is 100%; however, the 5-year survival rate for patients with metastatic prostate cancer is about 30%. These numbers translate to approximately 30,000 prostate cancer-related deaths every year1. It is now appreciated that patients may have indolent, intermediate, or aggressive disease, and it can be difficult to determine how to treat them. For patients with indolent disease, it may be harmful to perform interventions such as radical surgery, radiation, and chemotherapy. For those with aggressive disease, it is harmful to wait to treat with the same interventions because these cancers can progress relatively quickly. The prostate-specific antigen (PSA) blood test, combined with the digital rectal exam, histology from biopsies, and imaging (e.g. bone scans) give clinicians information about cancers to assist in making treatment decisions. Unfortunately, patients are still being over-treated, and men are still dying from prostate cancer due to a lack of more detailed prognostic tests and effective drugs to treat the appropriate disease state. Metastatic prostate cancer, on the whole, remains incurable.
These gaps in knowledge require experimental models with which to advance knowledge and discover and test novel therapeutics. The mouse, Mus musculus, has been used extensively to model prostate cancer and to test novel drugs preclinically. The mouse is the single most important mammal with which we can conduct experiments to further our knowledge of cancer biology2, 3. Mice are used in drug discovery to discover novel molecules involved in cancer, to determine whether cancer-associated molecules truly recapitulate the associated cancer property, and to test whether novel drugs are non-toxic and work to effectively combat cancer. The mouse has many advantages as a cancer model, and its use has led to many new discoveries. The mouse also has several disadvantages that may explain why so few drugs that work in mice do not work in humans. A lack of thorough understanding of how to use the mouse as a model may also hurt our collective ability to translate novel discoveries into novel drugs. In this review, we will discuss the advantages and disadvantages of using mice to model prostate cancer, the various types of models that have been used to do so, and we will end by providing our opinions as to the mouse’s utility and the future of mouse models in prostate cancer.
2.1 Mice and men: how similar are the organisms?
How similar mice are to men is an important question. Men and mice have some obvious similarities, such as both being mammalian. There are also some obvious differences, in that the mouse is approximately 300 times smaller than the man, walks on all fours, and has a tail. There are many more subtle characteristics by which we can compare the two. The mouse genome (specifically the C57BL/6 strain) was sequenced in 2002 – one year after the human genome was completed. The human genome is approximately 3.3 gigabases (Gb) in size, whereas the mouse genome is 2.7 Gb. Approximately 40% of the human genome can be aligned to the mouse genome, and over 90% of the two genomic regions have conserved synteny4. About 99% of mouse genes have a human ortholog, and the nucleotide and amino acid sequences of these orthologs are approximately 85% identical3, 5. There are of course differences between the sequences – telomere length (much longer in mouse)6, 7; GC content (42% in mouse, 41% in human); and activity of transposable elements (more active in the mouse lineage), for example4.
Further genetic analysis was completed in 2014, when six seminal papers were published comparing the human and mouse genomes and transcriptomes, as well as regulatory mechanisms8–13. It was found that while many genes share similar expression patterns, 4,767 genes were differentially expressed at a statistically significant level (2,569 were more highly expressed in humans, and 2,198 were more highly expressed in mice)12. An example of differential expression is in hematopoietic stem cells (HSCs), which express the cell surface marker CD34 at a high level in humans, while expressing CD38 at a low level. Conversely, mouse HSCs express high levels of CD38 and low levels of CD3414. The data also point to a high degree of divergence for sequences that are related to cis-transcriptional regulation and higher order chromatin organization11. In any given experiment, genomic and gene expression differences must be taken into consideration when performing any experiments involving mice, and care and caution must be taken in designing any experiment.
2.2 Mice and men: how similar are the prostates?
There are noteworthy differences between mouse and human prostate expression signatures. While mouse prostate cells respond to androgen stimulation and signaling similarly to humans15, 16, mice do not express certain human androgen-responsive genes, such as prostate-specific antigen (PSA) and prostate-specific membrane antigen (PSMA)17, 18. More broadly, there are other genes that are differentially expressed but many that are similarly expressed between mice and human in normal prostate tissue19 and prostate cancer20–22.
Mouse and human prostates differ anatomically. Human prostates are walnut-shaped and -sized, consisting of a single lobe and three zones (central, peripheral, and transitional) surrounding the urethra at the base of the bladder. The mouse prostate also surrounds the urethra at the base of the bladder, but is comprised of three pairs of lobes (anterior, ventral, and dorsolateral) one of which (anterior) protrude out immediately adjacent to the seminal vesicles (see Figure). Testosterone levels also differ – total and free plasma testosterone levels may vary between individual mice by 30-fold (likely due to episodic secretion23) and vary greatly between genetic backgrounds24. In addition, the average testosterone level of a hormonally intact mouse resembles hypogonadal human levels25. Adding and removing testosterone from mice drastically affects prostate tumor growth in xenograft models25.
Figure. Diagram of mouse and human prostate anatomy and histology.

Only one of each lobe is represented in the mouse prostate anatomy diagram. Protein markers that are commonly associated with each type of prostate cell are listed beneath the cell types. Adapted from references162, 163.
Histologically, the mouse prostate is strikingly similar to the human prostate, and great care has been placed into defining and describing the characteristics of normal and neoplastic prostate histology26. Mice differ in the ratio of basal cells to luminal cells, which is roughly 1:1 in humans but is 1 basal cell to 4 luminal cells in mice (see Figure)27. Importantly, mice rarely, if ever, spontaneously get prostate cancer – in a study of 612 wild-type mice, none were found to have prostate carcinoma28. This makes syngeneic models more difficult to generate and extremely rare, but the advantage is that the prostate cancer seen in mouse models is almost definitely caused by the experimental conditions, rather than by chance. Overall, the mouse prostate has many anatomic, histologic, and genetic similarities to the human and is a valuable mammalian organism with which to model human disease, as long as the differences are understood and taken into consideration.
2.3 Mouse models of prostate cancer
There are four main types of mouse models of prostate cancer: genetically engineered mouse models (GEMMs), cell line xenografts, patient-derived xenografts (PDXs), and syngeneic models. Each type has its own advantages and disadvantages (Table 1).
Table 1.
Types of cancer mouse models
| Model Type | Brief description | Advantages | Disadvantages |
|---|---|---|---|
| Genetically engineered mouse model (GEMM) | Target genes are knocked in or out in mouse cells (either whole body or tissue-specific), causing a phenotype |
|
|
| Cell line xenograft | Human (or other) cell lines are injected into immune-deficient mice |
|
|
| Patient- derived xenograft (PDX) | Human (or other) tumor tissue removed from patient is injected into immune-deficient mice |
|
|
| Syngeneic model | Mouse tumor tissue is injected into immune-competent mice of the same genetic background |
|
|
2.3a GEMMs
GEMMs can be broken down into three further categories: transgenics, whole-body knockouts, and conditional knockouts29. Transgenics use tissue-specific promoters to cause expression of a DNA construct. Whole body knockouts delete genes ubiquitously via homologous recombination in embryonic mouse stem cells30. To avoid embryonic lethality and whole-body deletion, conditional knockouts were developed, in which genes are deleted in a tissue-specific manner, most often using the Cre-loxP system29. Tissue-specific promoters drive expression of Cre recombinase, which then deletes any DNA flanked by loxP sequences31. Many prostate cancer GEMMs have been generated over the years (Table 2)21, 32–89.
Table 2.
Genetically engineered mouse models of prostate cancer
| Hyperplasia or prostatic intraepithelial neoplasia (PIN) | Carcinoma, no metastasis reported | Carcinoma, metastasis reported |
|---|---|---|
| p27 KO [50, 58, 70] Mt-PRL [84] Pten+/− [45] Nkx3.1 KO [35] PB-mAR [77] PSA-Cre;Nkx3.1-flox [33] PB-FGF8b [76] PB-Akt [67] PB-FGFR1 [52] MMTV-Cre;LSL-Catnblox(ex3) [37] MMTV-Cre;Pten-flox [34] FSP1-Cre;TGFβ-flox [36] PB-Cre;Rb-flox [65] PB-Ras [75] PB-Cre;LSL-Ph15LO-1 [56] PB-Cre;IGF1-flox [78] PB-Erg [60] Pten × p53 KO [43] PB-Cre;LSL-Catnblox(ex3) [85] PB-Cre;Brca2-flox;p53-flox [51] PB-PI3K p110B [61] PB-Cre;Pten-flox;CAG-Sox9 [79] PB-Cre;Scrib-flox [72] Nkx3.1-CreERT;Apc-flox [80] |
BK5-IGF1 [46] Pten × p27 KO [44] PB-myc [21] PB-Cre;Pten-flox;p53-flox [42] PB-neu [62] PB-Cre;Apc-flox [38] PB-Vav3 [63] PB-Akt × p27 KO [66] Pten+/− × Par4 KO [49] PB-Cre;LSL-Catnblox(ex3);LSL-KrasV12 [73] Nkx3.1-CreERT;Pten-flox [83] PB-Erg × Pten+/− [40] PB-hepsin;PB-myc [71] Pten+/− × Phlpp1 KO [41] PB-Cre;Scrib-flox;LSL-KrasG12D [72] PB-Cre;Pten-flox;p53-flox;LSL-cMyc [57] PB-Cre;p53-flox;Smad4-flox [48] PB-FGFR1 × H2Kb-iLrp5 [39] |
C3(1)-Tag [68] * PB-Tag (TRAMP) [55] * FG-Tag [74] * LPB-Tag (LADY) [89] * CR2-Tag [54] PSP94-Tag [53] LPB-Tag;PB-hepsin [59] Pten × Nkx3.1 KO [32] PSA-Cre;Pten-flox [64] PB-Cre;Pten-flox [82] PB-Cre;p53-flox;Rb-flox [87] PB-FGF8b × Pten+/− [86] PB-Cre;Pten-flox;Smad4-flox [47] * PB-Cre;Pten-flox;p53-flox;Smad4-flox [48] * PB-Cre;Pten-flox;p53-flox;LSL-Tert [48] PB-Cre;Pten-flox;LSL-KrasG12D [69] Nkx3.1-CreERT;Pten-flox;LSL-BrafV600E [81] |
Bone metastasis reported
Reference numbers in brackets [ ].
A drawback to using GEMMs is the length of time it takes to make them; it can take 6-12 months or longer to generate a model (depending on the number of genes being excised), and this is accompanied by the high cost of maintaining mouse colonies. New technology, the CRISPR-Cas9 system, has significantly reduced the time and cost, and it now can take a mere 4 weeks to generate a GEMM90. But even with a shorter generation time, genetically engineered mice are typically aged out a long time relative to xenograft models. Another issue with GEMMs is that they rarely metastasize to bone, which is a much-desired aspect of a prostate cancer model, although some models have reported bone metastases48. Even when metastases are present, mouse tumor cells are technically difficult to detect in normal mouse tissue, and the field is generally critical of reports of metastatic lesions as to whether or not the metastases are truly metastatic and are epithelial in origin91.
GEMMs have rarely been used for drug discovery. The TRAMP model (see Table 2) has been used to test antioxidants, anti-androgens, immune therapy, and other drugs92, 93. In another example, rapamycin (an mTOR inhibitor) and PD0325901 (a mitogen-activated protein kinase – MAPK – inhibitor) were tested on the Pten+/−; Nkx3.1ko mouse model32, 94. Two disadvantages to using a GEMM for drug discovery experiments such as these are that the mice often must be aged out for months before tumors consistently develop (this specific experiment waited 10 months), and that experimental groups are often smaller (n=4 in many of these experiments). The advantage of GEMMs is that they carry genetic modifications often associated with the human condition. Due to tissue-specific conditional models, these modifications can be made directly in prostate tissue in a cell-specific manner. This more closely resembles the human disease, in which mutations occur in the prostate cells themselves; this allows for a greater understanding of the molecular mechanistic causes of the disease. GEMMs are also useful for studying the stromal environment, as fibroblast-directed genetic modifications have been shown to drive prostate neoplasia36.
2.3b Cell line xenografts
In cell line xenograft models, human cancer cells are injected into immune-deficient mice. Due to the number of available cell lines, genes that can be manipulated, sites of injection, backgrounds of immune-deficient mice, and imaging modalities, there is a myriad of experiments that can be done using xenografts95. This model is used most often for drug studies, due to its reproducibility, quick experimental timeframe, and relative ease of experimental setup.
Several immune-deficient models exist: athymic nude96, severe-combined immunodeficiency (SCID)97, non-obese diabetic (NOD)-SCID98, NOD-SCID-interleukin 2 receptor gamma null (NSG)99, and recombination-activation gene (RAG)100, 101 (Table 3). Of these, the NSG mouse is the most immune-deficient, and has the highest take rate of human immune cells and cancer cells29, 99. The biggest issue with using immune-deficient mice for cancer research – even though they are required to grow human cancers – is that immune cells have been shown to play important roles in cancer progression and eradication102–104. The immune system’s roles in cancer therefore cannot be studied in immune-deficient mice. A final issue with this model is the relative lack of cell lines in prostate cancer105. While other types of cancer have dozens of cell lines to choose from, the vast majority of prostate cancer cell line work is done in PC3106, LNCaP and its derivatives107–109, VCaP110, MDA PCa111, and DU145112 lines.
Table 3.
Types of immune-deficient mice
| Type | Gene(s) affected | Immune cells that are deficient or defective |
|---|---|---|
| Athymic nude | Forkhead box N1 (Foxn1) | T cells |
| SCID | Protein kinase D1 (Prkd1) | T and B cells |
| NOD-SCID | Cytotoxic T-lymphocyte-associated protein 4 (Ctla4) & Prkd1 | T, B, NK, and complement cells |
| NSG | Interleukin 2 receptor gamma (Il2rg) & Prkd1 & Ctla4 | T, B, and NK cells, macrophages, and dendritic cells |
| RAG | Recombination activating genes 1 or 2 (Rag1 or Rag2) | T and B cells |
One potential way to study the immune system’s effect on cancer and metastasis in an immune-deficient model is to “humanize” the mouse by injecting human hematopoietic stem cells or differentiated immune cells, followed by injection of human cancer cells. These mice have no endogenous immune system but have some aspects of a human immune system within them. They have been studied primarily in the immunology field and for tumor vaccines, as well as in other fields113. The hu-PB-NOD/SCID model in particular has been used to study immune response to prostate cancer and to develop prostate cancer vaccines114–116. It would likely be beneficial to use these or similar mice not only for tumor immunology studies, but also for any xenograft study, as immune cells can affect tumor growth and metastasis. Some limitations of this model are incomplete hematopoietic differentiation, inadequate cellular interactions from perhaps under-developed immune cells, and limited production of human cytokines in mice113.
2.3c Patient-derived xenografts
A patient-derived xenograft (PDX) is generated through human tumor tissue being directly placed or injected into an immune-deficient mouse. This can be done using tissue obtained during surgery, or through needle biopsy at diagnosis117. Prostate tumors have been difficult to establish, as they require high quality tumor tissue and thorough characterization117–122. While PDX models capture more of the genomic, epigenetic, and proteomic diversity within prostate cancer, it has been difficult to use PDXs for drug discovery123. Even so, multiple institutions have now developed many novel PDXs, including the LuCaP models124–127, the bone metastatic BM18 model128, multiple models from MD Anderson129, 130, and others119, 131, 132. It is challenging to standardize therapeutic protocols since tumor tissue varies genetically and phenotypically between patients. It is believed, however, that PDXs represent the future of personalized or individualized medicine – each patient will some day be able to have their tumor grown as a PDX (or avatar) and analyzed for genomic alterations, identifying actionable targets for therapy123. Lessons learned from the many existing and ever-increasing number of PDXs should facilitate the harnessing of this technology for novel biological discoveries at each stage of prostate cancer122.
2.3d Syngeneic models
In a syngeneic model, tumor tissue from one mouse is removed and placed into another mouse of similar genetic background and therefore is immunologically compatible. The major upside to this model is that the immune system remains intact, which solves the problem of using immune-compromised mice in xenografts. With these models, tumors can be grown relatively quickly, which avoids the long timeline issue in GEMMs. However, syngeneic prostate cancer models are extremely rare due to the scarcity of mouse prostate tumors and cancer cell lines, and lack of effort being placed into generating immortal cell lines from mouse prostate cancer models. Some murine prostate cancer cell lines have been generated133–135, and they provide new opportunities for novel drug discovery in mice with intact immune systems. A syngeneic PTEN-deficient model was used to develop and test novel tumor vaccines, using murine cell lines PTEN-P8 and PTEN-CaP8, derived from a PTEN knockout model. These cells were used to develop the vaccine, and then injected into wild-type C57BL/6 mice, where the vaccine was tested136.
2.4 Using the mouse to test drugs
GEMMs are rarely used in drug studies, due to their lengthy time course, but they can be powerful models92. PDX and syngeneic models are also less often used for drug studies, although PDXs are becoming more popular. Syngeneic models are most often used in immunotherapy experiments due to the immune competency of the mice137–140. Human cell line xenograft models are by far the most often used for drug studies. The most common method to test drugs currently is to subcutaneously inject prostate cancer cells, treat with drug(s), and then measure the change in subcutaneous tumor volume over time. Depending on the cell line involved, the subcutaneous model may also be used to study metastasis and a drug’s effect on metastatic ability141.
Over the years, hundreds of drugs have been tested in mice to determine their efficacy in reducing tumor volume. Very few of these, however, have been translated into a drug to treat human cancer. In fact, only about 5% of anticancer compounds tested in animal models also show sufficient anticancer activity in phase III clinical trials and are ultimately approved by the FDA142. There are many reasons that could explain why tumors shrink in mice, but not in humans, using the same drug.
Subcutaneous cell line xenograft tumors grow in a foreign microenvironment with different host stroma than their native environment. Orthotopic injection models, in which prostate cancer cells are injected directly into the murine prostate, may better recapitulate the primary tumor’s microenvironment143–147, but are not as easy as the subcutaneous model to assess for growth and therapeutic effects.
The reproducibility that makes the cell line xenograft model so attractive for experimentation may also be confounding drug discovery and prognostic value. Lack of tumor heterogeneity may play a negative role in the ability to predict a drug’s effectiveness in humans117. A drug that may cause a clonal cell line tumor to shrink may have little to no effect on human prostate tumors that are extremely heterogeneous.
Mice are approximately 300 times smaller than humans, resulting in different pharmacokinetics and pharmacodynamics of drugs. A mouse’s average heart rate is approximately 600 beats per minute as compared to humans, whose average heart rate is 70-80 beats per minute. Mice in general can tolerate larger doses of drug because they can clear the chemicals from their system more quickly148. In general, however, allometric scaling laws are believed to work to extrapolate mouse drug concentrations to humans149.
The delivery mechanism of drugs may differ between mice and humans, depending on the drug’s solubility, mechanism of action, and metabolism. Drugs in humans are generally either oral or intravenous. Administration of oral drugs is technically difficult in mice, making translation to humans challenging. Drugs are commonly given to mice via intraperitoneal injections, which mimics intravenous administration in patients. Drug metabolism can also vary due to the expression of different enzymes between species. The drug abiraterone, for example, varies in tissue-specific expression of 17α hydroxylase (CYP17A1): it is expressed in the adrenal cortex as well as the gonads in humans, but only in the gonads in mice150. This expression difference should be kept in mind, but research has still been done with abiraterone in xenograft models151, 152.
There continues to be a notable lack of metastatic models in the prostate cancer field. Researchers often use pseudo-metastatic models, where cancer cells are injected directly into the heart or into the site of metastasis itself. These types of models bypass steps in the metastatic cascade, and therefore, drugs that are tested in these models are being measured against a simulated metastatic lesion, which is not representative of the metastases seen in human patients. Metastatic lesions are rarely found in GEMMs, possibly because metastases never actually develop, possibly because the primary tumor grows too quickly and the mice must be euthanized prior to development of metastases, or potentially because of immunosurveillance mechanisms. Metastases can be detected in xenograft models, however, and researchers should know which injection type to use, depending on the cell line95.
Some of the differences seen in drug responses between mice and men may not be due to the differences between the species at all but may be due to the poor experimental design. Some clinical trials do not have enough statistical power to find the true benefit of a drug that showed promise in mice153. Some clinical trials may give a drug later in a time course than was helpful in mice, thereby trying to have an effect too late. These two issues – not enough statistical power and bad timing – also plague the mouse studies as well. Other issues arise with experimental planning. There is innate bias in the interpretation of data that can be reduced with some quality control measures, such as blinding the study participants, randomizing the mice (or patients), and performing a priori statistical calculations to determine how many subjects are required. A study of 2,000 articles published between 1980 and 2000 in seven of the leading scientific journals (including Science, Nature, and Cell) discovered that very few studies actually use these quality control techniques154. Guidelines on how to care for and utilize mice in cancer research settings have been published, however, which has led to better practices in animal research155. Another study reviewed the animal model publications of stroke and concluded that a publication bias exists in which negative data is rarely published, and published articles generally report significant findings (a.k.a. positive data)156. This bias may skew the way the field interprets the value of a novel drug or molecule. The number of variables to take into account when designing a preclinical trial in mice is underappreciated157. Several recommendations have been published to improve the quality of preclinical research publications, including allowing for publication of negative data, increasing communication between physicians and scientists and patients, and increasing access to cancer research tools (such as mouse models) among research scientists158. Table 4 discusses many practical questions to consider when planning a mouse project.
Table 4.
Practical questions to consider when planning a mouse experiment
|
|
3. Conclusion
Metastatic prostate cancer is incurable. Experimental models are essential to discover novel therapeutic drug targets. The mouse is the best model organism for this purpose. On the organismal and prostate scales, mice have many similarities to humans. Of course, many differences also exist. These differences must be taken into consideration during the planning stages of any mouse experiment. Some experiments are not possible due to these differences, particularly in the realm of immunity. Shrewd planning and the development of innovative novel techniques have overcome many of these differences. Unfortunately, many new drugs work to shrink tumors in mice, but fail in human clinical trials. Many variables exist that could explain this phenomenon. Better planning, more quality control measures, and new technologies should help to increase the number of drugs that are translatable from mice to men.
4. Expert Opinion
The ultimate goal in prostate cancer research is to identify the underlying cellular and molecular causes of tumorigenesis, progression, castration resistance, and metastasis so that drugs can be developed to cure the specific stages of the disease. The mouse is not a perfect organism with which to complete this goal – however, a perfect model does not exist. Due to the numerous legitimate ethical issues with performing research on humans, model organisms such as the mouse will be necessary until better models can be developed.
The attrition rates between the discovery of a successful therapeutic agent in a mouse study and approval for human use remains appalling. The field suffers from use of clonal cell lines, lack of immunocompetent models that recapitulate human disease, lack of understanding of how mice metabolize agents, and lack of well-designed rigorous clinical trials in mice105. The situation need not be as dire as it sounds. Though there is not a single set of instructions for every mouse experiment to work perfectly, there are certain rules, guidelines, and sensible practices that if followed would increase the probability of translatable data to be produced and bias to be reduced (Table 4 and ref. 157).
Perhaps the biggest challenge in the prostate cancer field is the generation of an immune-competent mouse model that recapitulates the late stages of prostate cancer, particularly bone metastasis. Bone metastasis is what is killing prostate cancer patients, but we still lack experimental models with which to discover new molecules and drugs. Some GEMMs have reported bone metastasis, albeit infrequently, and these metastatic lesions should be validated and turned into cell lines that can be used for syngeneic models. Immune-deficient mice with a “humanized” immune system might also be useful in this purpose. These types of models hold much potential.
Several areas of research hold particular promise in the coming years. As the number of PDX models increases, there should be a wealth of information representing an ever-expanding population of prostate tumors. Though reproducibility will likely remain low due to the heterogeneous nature of prostate cancer, these experimental models are the most realistic of human cancer and should provide new directions and molecular strategies for therapeutics. The CRISPR/Cas9 genomic modulation system holds incredible potential for swiftly advancing the number of GEMMs, thereby allowing for swifter validation of candidate genes as tumorigenic or metastatic. The decreased time and cost to make a conditional knockout or mice expressing a gene with a point mutation allows for much more flexibility than previous years and opens the doors to many more possibilities. Finally, as imaging technologies improve, in vivo detection of tumor size, disseminated tumor cells, and metastatic lesions will vastly improve our ability to test drugs in an efficient manner over longer time courses159–161.
The future of prostate cancer research using mouse models is moving in the right direction. We are constantly gaining new knowledge and insight and are learning from past mistakes122, 158. Hopefully, researchers will respect the differences that exist between mice and men and will move with care and caution, ensuring that our work is performed and recorded with quality control measures in place. The increasing number and quality of open access journals should also improve the amount of “negative” data available, so as to open up further lines of communication to determine the true value of a molecule or compound being used in anti-cancer studies. Free-flowing communication and information will be the key for the ultimate goal to be realized.
Highlights.
Metastatic prostate cancer is incurable, and indolent prostate cancer is over-treated. More experimental models are needed, and the mouse is an ideal model organism.
Mice and men have many similarities at the genomic and anatomic level, but also many differences. These differences should be considered when planning, interpreting, and reporting results.
Four types of prostate cancer models exist: genetically engineered mouse models (GEMMs), cell line xenografts, patient-derived xenograft (PDXs), and syngeneic models.
Very few drug studies conducted in mouse models have produced effective drugs for human use.
Mouse-human biological differences, tumor stroma differences, lack of tumor heterogeneity and immune cells in mouse models, poor clinical trial design, and publication bias could explain this attrition rate.
Bibliography
- 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA: a cancer journal for clinicians. 2015 Jan-Feb;65(1):5–29. doi: 10.3322/caac.21254. [DOI] [PubMed] [Google Scholar]
- 2.Paigen K. A miracle enough: the power of mice. Nature medicine. 1995 Mar;1(3):215–20. doi: 10.1038/nm0395-215. [DOI] [PubMed] [Google Scholar]
- 3.Guenet JL. The mouse genome. Genome research. 2005 Dec;15(12):1729–40. doi: 10.1101/gr.3728305. [DOI] [PubMed] [Google Scholar]
- 4**.Mouse Genome Sequencing C. Waterston RH, Lindblad-Toh K, Birney E, Rogers J, Abril JF, et al. Initial sequencing and comparative analysis of the mouse genome. Nature. 2002 Dec 5;420(6915):520–62. doi: 10.1038/nature01262. First article to compare genomic sequence between mice and humans. [DOI] [PubMed] [Google Scholar]
- 5.Makalowski W, Zhang J, Boguski MS. Comparative analysis of 1196 orthologous mouse and human full-length mRNA and protein sequences. Genome research. 1996 Sep;6(9):846–57. doi: 10.1101/gr.6.9.846. [DOI] [PubMed] [Google Scholar]
- 6.Broccoli D, Godley LA, Donehower LA, Varmus HE, de Lange T. Telomerase activation in mouse mammary tumors: lack of detectable telomere shortening and evidence for regulation of telomerase RNA with cell proliferation. Molecular and cellular biology. 1996 Jul;16(7):3765–72. doi: 10.1128/mcb.16.7.3765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kipling D, Cooke HJ. Hypervariable ultra-long telomeres in mice. Nature. 1990 Sep 27;347(6291):400–2. doi: 10.1038/347400a0. [DOI] [PubMed] [Google Scholar]
- 8.Cheng Y, Ma Z, Kim BH, Wu W, Cayting P, Boyle AP, et al. Principles of regulatory information conservation between mouse and human. Nature. 2014 Nov 20;515(7527):371–5. doi: 10.1038/nature13985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pope BD, Ryba T, Dileep V, Yue F, Wu W, Denas O, et al. Topologically associating domains are stable units of replication-timing regulation. Nature. 2014 Nov 20;515(7527):402–5. doi: 10.1038/nature13986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Stergachis AB, Neph S, Sandstrom R, Haugen E, Reynolds AP, Zhang M, et al. Conservation of trans-acting circuitry during mammalian regulatory evolution. Nature. 2014 Nov 20;515(7527):365–70. doi: 10.1038/nature13972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yue F, Cheng Y, Breschi A, Vierstra J, Wu W, Ryba T, et al. A comparative encyclopedia of DNA elements in the mouse genome. Nature. 2014 Nov 20;515(7527):355–64. doi: 10.1038/nature13992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12**.Lin S, Lin Y, Nery JR, Urich MA, Breschi A, Davis CA, et al. Comparison of the transcriptional landscapes between human and mouse tissues. Proceedings of the National Academy of Sciences of the United States of America. 2014 Dec 2;111(48):17224–9. doi: 10.1073/pnas.1413624111. Part of a seminal series in 2014 comparing mice and humans at the genomic and gene expression levels. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vierstra J, Rynes E, Sandstrom R, Zhang M, Canfield T, Hansen RS, et al. Mouse regulatory DNA landscapes reveal global principles of cis-regulatory evolution. Science. 2014 Nov 21;346(6212):1007–12. doi: 10.1126/science.1246426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Challen GA, Boles N, Lin KK, Goodell MA. Mouse hematopoietic stem cell identification and analysis. Cytometry Part A : the journal of the International Society for Analytical Cytology. 2009 Jan;75(1):14–24. doi: 10.1002/cyto.a.20674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Donjacour AA, Cunha GR. The effect of androgen deprivation on branching morphogenesis in the mouse prostate. Developmental biology. 1988 Jul;128(1):1–14. doi: 10.1016/0012-1606(88)90260-6. [DOI] [PubMed] [Google Scholar]
- 16.Hsieh CL, Xie Z, Liu ZY, Green JE, Martin WD, Datta MW, et al. A luciferase transgenic mouse model: visualization of prostate development and its androgen responsiveness in live animals. Journal of molecular endocrinology. 2005 Oct;35(2):293–304. doi: 10.1677/jme.1.01722. [DOI] [PubMed] [Google Scholar]
- 17.Aggarwal S, Ricklis RM, Williams SA, Denmeade SR. Comparative study of PSMA expression in the prostate of mouse, dog, monkey, and human. The Prostate. 2006 Jun 15;66(9):903–10. doi: 10.1002/pros.20413. [DOI] [PubMed] [Google Scholar]
- 18.Wei C, Willis RA, Tilton BR, Looney RJ, Lord EM, Barth RK, et al. Tissue-specific expression of the human prostate-specific antigen gene in transgenic mice: implications for tolerance and immunotherapy. Proceedings of the National Academy of Sciences of the United States of America. 1997 Jun 10;94(12):6369–74. doi: 10.1073/pnas.94.12.6369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Berquin IM, Min Y, Wu R, Wu H, Chen YQ. Expression signature of the mouse prostate. The Journal of biological chemistry. 2005 Oct 28;280(43):36442–51. doi: 10.1074/jbc.M504945200. [DOI] [PubMed] [Google Scholar]
- 20.Calvo A, Xiao N, Kang J, Best CJ, Leiva I, Emmert-Buck MR, et al. Alterations in gene expression profiles during prostate cancer progression: functional correlations to tumorigenicity and down-regulation of selenoprotein-P in mouse and human tumors. Cancer research. 2002 Sep 15;62(18):5325–35. [PubMed] [Google Scholar]
- 21.Ellwood-Yen K, Graeber TG, Wongvipat J, Iruela-Arispe ML, Zhang J, Matusik R, et al. Myc-driven murine prostate cancer shares molecular features with human prostate tumors. Cancer cell. 2003 Sep;4(3):223–38. doi: 10.1016/s1535-6108(03)00197-1. [DOI] [PubMed] [Google Scholar]
- 22.Haram KM, Peltier HJ, Lu B, Bhasin M, Otu HH, Choy B, et al. Gene expression profile of mouse prostate tumors reveals dysregulations in major biological processes and identifies potential murine targets for preclinical development of human prostate cancer therapy. The Prostate. 2008 Oct 1;68(14):1517–30. doi: 10.1002/pros.20803. [DOI] [PubMed] [Google Scholar]
- 23.Bartke A, Dalterio S. Evidence for episodic secretion of testosterone in laboratory mice. Steroids. 1975 Dec;26(6):749–56. doi: 10.1016/0039-128x(75)90107-5. [DOI] [PubMed] [Google Scholar]
- 24.Bartke A, Steele RE, Musto N, Caldwell BV. Fluctuations in plasma testosterone levels in adult male rats and mice. Endocrinology. 1973 Apr;92(4):1223–8. doi: 10.1210/endo-92-4-1223. [DOI] [PubMed] [Google Scholar]
- 25.Michiel Sedelaar JP, Dalrymple SS, Isaacs JT. Of mice and men--warning: intact versus castrated adult male mice as xenograft hosts are equivalent to hypogonadal versus abiraterone treated aging human males, respectively. The Prostate. 2013 Sep;73(12):1316–25. doi: 10.1002/pros.22677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26*.Shappell SB, Thomas GV, Roberts RL, Herbert R, Ittmann MM, Rubin MA, et al. Prostate pathology of genetically engineered mice: definitions and classification. The consensus report from the Bar Harbor meeting of the Mouse Models of Human Cancer Consortium Prostate Pathology Committee. Cancer research. 2004 Mar 15;64(6):2270–305. doi: 10.1158/0008-5472.can-03-0946. Seminal paper discussing the pathologic definitions of mouse prostate histology, compared to human prostate histology. [DOI] [PubMed] [Google Scholar]
- 27.El-Alfy M, Pelletier G, Hermo LS, Labrie F. Unique features of the basal cells of human prostate epithelium. Microscopy research and technique. 2000 Dec 1;51(5):436–46. doi: 10.1002/1097-0029(20001201)51:5<436::AID-JEMT6>3.0.CO;2-T. [DOI] [PubMed] [Google Scholar]
- 28.Suwa T, Nyska A, Haseman JK, Mahler JF, Maronpot RR. Spontaneous lesions in control B6C3F1 mice and recommended sectioning of male accessory sex organs. Toxicologic pathology. 2002 Mar-Apr;30(2):228–34. doi: 10.1080/019262302753559560. [DOI] [PubMed] [Google Scholar]
- 29*.Valkenburg KC, Williams BO. Mouse models of prostate cancer. Prostate cancer. 2011;2011:895238. doi: 10.1155/2011/895238. Discusses many mouse models of prostate cancer, particularly GEMMs, up to the year 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Capecchi MR. Altering the genome by homologous recombination. Science. 1989 Jun 16;244(4910):1288–92. doi: 10.1126/science.2660260. [DOI] [PubMed] [Google Scholar]
- 31.Sauer B, Henderson N. Cre-stimulated recombination at loxP-containing DNA sequences placed into the mammalian genome. Nucleic acids research. 1989 Jan 11;17(1):147–61. doi: 10.1093/nar/17.1.147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Abate-Shen C, Banach-Petrosky WA, Sun X, Economides KD, Desai N, Gregg JP, et al. Nkx3.1; Pten mutant mice develop invasive prostate adenocarcinoma and lymph node metastases. Cancer research. 2003 Jul 15;63(14):3886–90. [PubMed] [Google Scholar]
- 33.Abdulkadir SA, Magee JA, Peters TJ, Kaleem Z, Naughton CK, Humphrey PA, et al. Conditional loss of Nkx3.1 in adult mice induces prostatic intraepithelial neoplasia. Molecular and cellular biology. 2002 Mar;22(5):1495–503. doi: 10.1128/mcb.22.5.1495-1503.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Backman SA, Ghazarian D, So K, Sanchez O, Wagner KU, Hennighausen L, et al. Early onset of neoplasia in the prostate and skin of mice with tissue-specific deletion of Pten. Proceedings of the National Academy of Sciences of the United States of America. 2004 Feb 10;101(6):1725–30. doi: 10.1073/pnas.0308217100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bhatia-Gaur R, Donjacour AA, Sciavolino PJ, Kim M, Desai N, Young P, et al. Roles for Nkx3.1 in prostate development and cancer. Genes & development. 1999 Apr 15;13(8):966–77. doi: 10.1101/gad.13.8.966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004 Feb 6;303(5659):848–51. doi: 10.1126/science.1090922. [DOI] [PubMed] [Google Scholar]
- 37.Bierie B, Nozawa M, Renou JP, Shillingford JM, Morgan F, Oka T, et al. Activation of beta-catenin in prostate epithelium induces hyperplasias and squamous transdifferentiation. Oncogene. 2003 Jun 19;22(25):3875–87. doi: 10.1038/sj.onc.1206426. [DOI] [PubMed] [Google Scholar]
- 38.Bruxvoort KJ, Charbonneau HM, Giambernardi TA, Goolsby JC, Qian CN, Zylstra CR, et al. Inactivation of Apc in the mouse prostate causes prostate carcinoma. Cancer research. 2007 Mar 15;67(6):2490–6. doi: 10.1158/0008-5472.CAN-06-3028. [DOI] [PubMed] [Google Scholar]
- 39.Carstens JL, Shahi P, Van Tsang S, Smith B, Creighton CJ, Zhang Y, et al. FGFR1-WNT-TGF-beta signaling in prostate cancer mouse models recapitulates human reactive stroma. Cancer research. 2014 Jan 15;74(2):609–20. doi: 10.1158/0008-5472.CAN-13-1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Carver BS, Tran J, Gopalan A, Chen Z, Shaikh S, Carracedo A, et al. Aberrant ERG expression cooperates with loss of PTEN to promote cancer progression in the prostate. Nature genetics. 2009 May;41(5):619–24. doi: 10.1038/ng.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen M, Pratt CP, Zeeman ME, Schultz N, Taylor BS, O’Neill A, et al. Identification of PHLPP1 as a tumor suppressor reveals the role of feedback activation in PTEN-mutant prostate cancer progression. Cancer cell. 2011 Aug 16;20(2):173–86. doi: 10.1016/j.ccr.2011.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Chen Z, Trotman LC, Shaffer D, Lin HK, Dotan ZA, Niki M, et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature. 2005 Aug 4;436(7051):725–30. doi: 10.1038/nature03918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Couto SS, Cao M, Duarte PC, Banach-Petrosky W, Wang S, Romanienko P, et al. Simultaneous haploinsufficiency of Pten and Trp53 tumor suppressor genes accelerates tumorigenesis in a mouse model of prostate cancer. Differentiation; research in biological diversity. 2009 Jan;77(1):103–11. doi: 10.1016/j.diff.2008.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Di Cristofano A, De Acetis M, Koff A, Cordon-Cardo C, Pandolfi PP. Pten and p27KIP1 cooperate in prostate cancer tumor suppression in the mouse. Nature genetics. 2001 Feb;27(2):222–4. doi: 10.1038/84879. [DOI] [PubMed] [Google Scholar]
- 45.Di Cristofano A, Pesce B, Cordon-Cardo C, Pandolfi PP. Pten is essential for embryonic development and tumour suppression. Nature genetics. 1998 Aug;19(4):348–55. doi: 10.1038/1235. [DOI] [PubMed] [Google Scholar]
- 46.DiGiovanni J, Kiguchi K, Frijhoff A, Wilker E, Bol DK, Beltran L, et al. Deregulated expression of insulin-like growth factor 1 in prostate epithelium leads to neoplasia in transgenic mice. Proceedings of the National Academy of Sciences of the United States of America. 2000 Mar 28;97(7):3455–60. doi: 10.1073/pnas.97.7.3455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ding Z, Wu CJ, Chu GC, Xiao Y, Ho D, Zhang J, et al. SMAD4-dependent barrier constrains prostate cancer growth and metastatic progression. Nature. 2011 Feb 10;470(7333):269–73. doi: 10.1038/nature09677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48**.Ding Z, Wu CJ, Jaskelioff M, Ivanova E, Kost-Alimova M, Protopopov A, et al. Telomerase reactivation following telomere dysfunction yields murine prostate tumors with bone metastases. Cell. 2012 Mar 2;148(5):896–907. doi: 10.1016/j.cell.2012.01.039. First prostate-specific knockout GEMM to report consistent bone metastasis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fernandez-Marcos PJ, Abu-Baker S, Joshi J, Galvez A, Castilla EA, Canamero M, et al. Simultaneous inactivation of Par-4 and PTEN in vivo leads to synergistic NF-kappaB activation and invasive prostate carcinoma. Proceedings of the National Academy of Sciences of the United States of America. 2009 Aug 4;106(31):12962–7. doi: 10.1073/pnas.0813055106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Fero ML, Rivkin M, Tasch M, Porter P, Carow CE, Firpo E, et al. A syndrome of multiorgan hyperplasia with features of gigantism, tumorigenesis, and female sterility in p27(Kip1)-deficient mice. Cell. 1996 May 31;85(5):733–44. doi: 10.1016/s0092-8674(00)81239-8. [DOI] [PubMed] [Google Scholar]
- 51.Francis JC, McCarthy A, Thomsen MK, Ashworth A, Swain A. Brca2 and Trp53 deficiency cooperate in the progression of mouse prostate tumourigenesis. PLoS genetics. 2010 Jun;6(6):e1000995. doi: 10.1371/journal.pgen.1000995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Freeman KW, Welm BE, Gangula RD, Rosen JM, Ittmann M, Greenberg NM, et al. Inducible prostate intraepithelial neoplasia with reversible hyperplasia in conditional FGFR1-expressing mice. Cancer research. 2003 Dec 1;63(23):8256–63. [PubMed] [Google Scholar]
- 53.Gabril MY, Onita T, Ji PG, Sakai H, Chan FL, Koropatnick J, et al. Prostate targeting: PSP94 gene promoter/enhancer region directed prostate tissue-specific expression in a transgenic mouse prostate cancer model. Gene therapy. 2002 Dec;9(23):1589–99. doi: 10.1038/sj.gt.3301895. [DOI] [PubMed] [Google Scholar]
- 54.Garabedian EM, Humphrey PA, Gordon JI. A transgenic mouse model of metastatic prostate cancer originating from neuroendocrine cells. Proceedings of the National Academy of Sciences of the United States of America. 1998 Dec 22;95(26):15382–7. doi: 10.1073/pnas.95.26.15382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Greenberg NM, DeMayo FJ, Sheppard PC, Barrios R, Lebovitz R, Finegold M, et al. The rat probasin gene promoter directs hormonally and developmentally regulated expression of a heterologous gene specifically to the prostate in transgenic mice. Molecular endocrinology. 1994 Feb;8(2):230–9. doi: 10.1210/mend.8.2.8170479. [DOI] [PubMed] [Google Scholar]
- 56.Kelavkar UP, Parwani AV, Shappell SB, Martin WD. Conditional expression of human 15-lipoxygenase-1 in mouse prostate induces prostatic intraepithelial neoplasia: the FLiMP mouse model. Neoplasia. 2006 Jun;8(6):510–22. doi: 10.1593/neo.06202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim J, Roh M, Doubinskaia I, Algarroba GN, Eltoum IE, Abdulkadir SA. A mouse model of heterogeneous, c-MYC-initiated prostate cancer with loss of Pten and p53. Oncogene. 2012 Jan 19;31(3):322–32. doi: 10.1038/onc.2011.236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kiyokawa H, Kineman RD, Manova-Todorova KO, Soares VC, Hoffman ES, Ono M, et al. Enhanced growth of mice lacking the cyclin-dependent kinase inhibitor function of p27(Kip1) Cell. 1996 May 31;85(5):721–32. doi: 10.1016/s0092-8674(00)81238-6. [DOI] [PubMed] [Google Scholar]
- 59.Klezovitch O, Chevillet J, Mirosevich J, Roberts RL, Matusik RJ, Vasioukhin V. Hepsin promotes prostate cancer progression and metastasis. Cancer cell. 2004 Aug;6(2):185–95. doi: 10.1016/j.ccr.2004.07.008. [DOI] [PubMed] [Google Scholar]
- 60.Klezovitch O, Risk M, Coleman I, Lucas JM, Null M, True LD, et al. A causal role for ERG in neoplastic transformation of prostate epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2008 Feb 12;105(6):2105–10. doi: 10.1073/pnas.0711711105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lee SH, Poulogiannis G, Pyne S, Jia S, Zou L, Signoretti S, et al. A constitutively activated form of the p110beta isoform of PI3-kinase induces prostatic intraepithelial neoplasia in mice. Proceedings of the National Academy of Sciences of the United States of America. 2010 Jun 15;107(24):11002–7. doi: 10.1073/pnas.1005642107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Li Z, Szabolcs M, Terwilliger JD, Efstratiadis A. Prostatic intraepithelial neoplasia and adenocarcinoma in mice expressing a probasin-Neu oncogenic transgene. Carcinogenesis. 2006 May;27(5):1054–67. doi: 10.1093/carcin/bgi324. [DOI] [PubMed] [Google Scholar]
- 63.Liu Y, Mo JQ, Hu Q, Boivin G, Levin L, Lu S, et al. Targeted overexpression of vav3 oncogene in prostatic epithelium induces nonbacterial prostatitis and prostate cancer. Cancer research. 2008 Aug 1;68(15):6396–406. doi: 10.1158/0008-5472.CAN-08-0645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Ma X, Ziel-van der Made AC, Autar B, van der Korput HA, Vermeij M, van Duijn P, et al. Targeted biallelic inactivation of Pten in the mouse prostate leads to prostate cancer accompanied by increased epithelial cell proliferation but not by reduced apoptosis. Cancer research. 2005 Jul 1;65(13):5730–9. doi: 10.1158/0008-5472.CAN-04-4519. [DOI] [PubMed] [Google Scholar]
- 65.Maddison LA, Sutherland BW, Barrios RJ, Greenberg NM. Conditional deletion of Rb causes early stage prostate cancer. Cancer research. 2004 Sep 1;64(17):6018–25. doi: 10.1158/0008-5472.CAN-03-2509. [DOI] [PubMed] [Google Scholar]
- 66.Majumder PK, Grisanzio C, O’Connell F, Barry M, Brito JM, Xu Q, et al. A prostatic intraepithelial neoplasia-dependent p27 Kip1 checkpoint induces senescence and inhibits cell proliferation and cancer progression. Cancer cell. 2008 Aug 12;14(2):146–55. doi: 10.1016/j.ccr.2008.06.00. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Majumder PK, Yeh JJ, George DJ, Febbo PG, Kum J, Xue Q, et al. Prostate intraepithelial neoplasia induced by prostate restricted Akt activation: the MPAKT model. Proceedings of the National Academy of Sciences of the United States of America. 2003 Jun 24;100(13):7841–6. doi: 10.1073/pnas.1232229100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Maroulakou IG, Anver M, Garrett L, Green JE. Prostate and mammary adenocarcinoma in transgenic mice carrying a rat C3(1) simian virus 40 large tumor antigen fusion gene. Proceedings of the National Academy of Sciences of the United States of America. 1994 Nov 8;91(23):11236–40. doi: 10.1073/pnas.91.23.11236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mulholland DJ, Kobayashi N, Ruscetti M, Zhi A, Tran LM, Huang J, et al. Pten loss and RAS/MAPK activation cooperate to promote EMT and metastasis initiated from prostate cancer stem/progenitor cells. Cancer research. 2012 Apr 1;72(7):1878–89. doi: 10.1158/0008-5472.CAN-11-3132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Nakayama K, Ishida N, Shirane M, Inomata A, Inoue T, Shishido N, et al. Mice lacking p27(Kip1) display increased body size, multiple organ hyperplasia, retinal dysplasia, and pituitary tumors. Cell. 1996 May 31;85(5):707–20. doi: 10.1016/s0092-8674(00)81237-4. [DOI] [PubMed] [Google Scholar]
- 71.Nandana S, Ellwood-Yen K, Sawyers C, Wills M, Weidow B, Case T, et al. Hepsin cooperates with MYC in the progression of adenocarcinoma in a prostate cancer mouse model. The Prostate. 2010 May 1;70(6):591–600. doi: 10.1002/pros.21093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pearson HB, Perez-Mancera PA, Dow LE, Ryan A, Tennstedt P, Bogani D, et al. SCRIB expression is deregulated in human prostate cancer, and its deficiency in mice promotes prostate neoplasia. The Journal of clinical investigation. 2011 Nov;121(11):4257–67. doi: 10.1172/JCI58509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Pearson HB, Phesse TJ, Clarke AR. K-ras and Wnt signaling synergize to accelerate prostate tumorigenesis in the mouse. Cancer research. 2009 Jan 1;69(1):94–101. doi: 10.1158/0008-5472.CAN-08-2895. [DOI] [PubMed] [Google Scholar]
- 74.Perez-Stable C, Altman NH, Brown J, Harbison M, Cray C, Roos BA. Prostate, adrenocortical, and brown adipose tumors in fetal globin/T antigen transgenic mice. Laboratory investigation; a journal of technical methods and pathology. 1996 Feb;74(2):363–73. [PubMed] [Google Scholar]
- 75.Scherl A, Li JF, Cardiff RD, Schreiber-Agus N. Prostatic intraepithelial neoplasia and intestinal metaplasia in prostates of probasin-RAS transgenic mice. The Prostate. 2004 Jun 1;59(4):448–59. doi: 10.1002/pros.20020. [DOI] [PubMed] [Google Scholar]
- 76.Song Z, Wu X, Powell WC, Cardiff RD, Cohen MB, Tin RT, et al. Fibroblast growth factor 8 isoform B overexpression in prostate epithelium: a new mouse model for prostatic intraepithelial neoplasia. Cancer research. 2002 Sep 1;62(17):5096–105. [PubMed] [Google Scholar]
- 77.Stanbrough M, Leav I, Kwan PW, Bubley GJ, Balk SP. Prostatic intraepithelial neoplasia in mice expressing an androgen receptor transgene in prostate epithelium. Proceedings of the National Academy of Sciences of the United States of America. 2001 Sep 11;98(19):10823–8. doi: 10.1073/pnas.191235898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sutherland BW, Knoblaugh SE, Kaplan-Lefko PJ, Wang F, Holzenberger M, Greenberg NM. Conditional deletion of insulin-like growth factor-I receptor in prostate epithelium. Cancer research. 2008 May 1;68(9):3495–504. doi: 10.1158/0008-5472.CAN-07-6531. [DOI] [PubMed] [Google Scholar]
- 79.Thomsen MK, Ambroisine L, Wynn S, Cheah KS, Foster CS, Fisher G, et al. SOX9 elevation in the prostate promotes proliferation and cooperates with PTEN loss to drive tumor formation. Cancer research. 2010 Feb 1;70(3):979–87. doi: 10.1158/0008-5472.CAN-09-2370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Valkenburg KC, Yu X, De Marzo AM, Spiering TJ, Matusik RJ, Williams BO. Activation of Wnt/beta-catenin signaling in a subpopulation of murine prostate luminal epithelial cells induces high grade prostate intraepithelial neoplasia. The Prostate. 2014 Nov;74(15):1506–20. doi: 10.1002/pros.22868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Wang J, Kobayashi T, Floc’h N, Kinkade CW, Aytes A, Dankort D, et al. B-Raf activation cooperates with PTEN loss to drive c-Myc expression in advanced prostate cancer. Cancer research. 2012 Sep 15;72(18):4765–76. doi: 10.1158/0008-5472.CAN-12-0820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wang S, Gao J, Lei Q, Rozengurt N, Pritchard C, Jiao J, et al. Prostate-specific deletion of the murine Pten tumor suppressor gene leads to metastatic prostate cancer. Cancer cell. 2003 Sep;4(3):209–21. doi: 10.1016/s1535-6108(03)00215-0. [DOI] [PubMed] [Google Scholar]
- 83.Wang X, Kruithof-de Julio M, Economides KD, Walker D, Yu H, Halili MV, et al. A luminal epithelial stem cell that is a cell of origin for prostate cancer. Nature. 2009 Sep 24;461(7263):495–500. doi: 10.1038/nature08361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wennbo H, Kindblom J, Isaksson OG, Tornell J. Transgenic mice overexpressing the prolactin gene develop dramatic enlargement of the prostate gland. Endocrinology. 1997 Oct;138(10):4410–5. doi: 10.1210/endo.138.10.5461. [DOI] [PubMed] [Google Scholar]
- 85.Yu X, Wang Y, Jiang M, Bierie B, Roy-Burman P, Shen MM, et al. Activation of beta-Catenin in mouse prostate causes HGPIN and continuous prostate growth after castration. The Prostate. 2009 Feb 15;69(3):249–62. doi: 10.1002/pros.20877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Zhong C, Saribekyan G, Liao CP, Cohen MB, Roy-Burman P. Cooperation between FGF8b overexpression and PTEN deficiency in prostate tumorigenesis. Cancer research. 2006 Feb 15;66(4):2188–94. doi: 10.1158/0008-5472.CAN-05-3440. [DOI] [PubMed] [Google Scholar]
- 87.Zhou Z, Flesken-Nikitin A, Corney DC, Wang W, Goodrich DW, Roy-Burman P, et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer research. 2006 Aug 15;66(16):7889–98. doi: 10.1158/0008-5472.CAN-06-0486. [DOI] [PubMed] [Google Scholar]
- 88.Yu X, Wang Y, DeGraff DJ, Wills ML, Matusik RJ. Wnt/beta-catenin activation promotes prostate tumor progression in a mouse model. Oncogene. 2011 Apr 21;30(16):1868–79. doi: 10.1038/onc.2010.560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Yan Y, Sheppard PC, Kasper S, Lin L, Hoare S, Kapoor A, et al. Large fragment of the probasin promoter targets high levels of transgene expression to the prostate of transgenic mice. The Prostate. 1997 Jul 1;32(2):129–39. doi: 10.1002/(sici)1097-0045(19970701)32:2<129::aid-pros8>3.0.co;2-h. [DOI] [PubMed] [Google Scholar]
- 90**.Wang H, Yang H, Shivalila CS, Dawlaty MM, Cheng AW, Zhang F, et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell. 2013 May 9;153(4):910–8. doi: 10.1016/j.cell.2013.04.025. Important paper discussing new methodology using the CRISPR/Cas9 system to make GEMMs more quickly and cost effectively. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Chiaverotti T, Couto SS, Donjacour A, Mao JH, Nagase H, Cardiff RD, et al. Dissociation of epithelial and neuroendocrine carcinoma lineages in the transgenic adenocarcinoma of mouse prostate model of prostate cancer. The American journal of pathology. 2008 Jan;172(1):236–46. doi: 10.2353/ajpath.2008.070602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ahmad I, Sansom OJ, Leung HY. The role of murine models of prostate cancer in drug target discovery and validation. Expert opinion on drug discovery. 2009 Aug;4(8):879–88. doi: 10.1517/17460440903049308. [DOI] [PubMed] [Google Scholar]
- 93.Kwon ED, Foster BA, Hurwitz AA, Madias C, Allison JP, Greenberg NM, et al. Elimination of residual metastatic prostate cancer after surgery and adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proceedings of the National Academy of Sciences of the United States of America. 1999 Dec 21;96(26):15074–9. doi: 10.1073/pnas.96.26.15074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94**.Kinkade CW, Castillo-Martin M, Puzio-Kuter A, Yan J, Foster TH, Gao H, et al. Targeting AKT/mTOR and ERK MAPK signaling inhibits hormone-refractory prostate cancer in a preclinical mouse model. The Journal of clinical investigation. 2008 Sep;118(9):3051–64. doi: 10.1172/JCI34764. A rare example of a GEMM being used for direct drug testing and discovery. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95*.Park SI, Kim SJ, McCauley LK, Gallick GE. Pre-clinical mouse models of human prostate cancer and their utility in drug discovery. Current protocols in pharmacology/editorial board, SJ Enna. 2010 Dec; doi: 10.1002/0471141755.ph1415s51. Chapter 14:Unit 14 15. Excellent protocol paper outlining the various injection sites for xenograft models. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Flanagan SP. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genetical research. 1966 Dec;8(3):295–309. doi: 10.1017/s0016672300010168. [DOI] [PubMed] [Google Scholar]
- 97.Bosma GC, Custer RP, Bosma MJ. A severe combined immunodeficiency mutation in the mouse. Nature. 1983 Feb 10;301(5900):527–30. doi: 10.1038/301527a0. [DOI] [PubMed] [Google Scholar]
- 98.Greiner DL, Shultz LD, Yates J, Appel MC, Perdrizet G, Hesselton RM, et al. Improved engraftment of human spleen cells in NOD/LtSz-scid/scid mice as compared with C.B-17-scid/scid mice. The American journal of pathology. 1995 Apr;146(4):888–902. [PMC free article] [PubMed] [Google Scholar]
- 99.Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K, et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002 Nov 1;100(9):3175–82. doi: 10.1182/blood-2001-12-0207. [DOI] [PubMed] [Google Scholar]
- 100.Mombaerts P, Iacomini J, Johnson RS, Herrup K, Tonegawa S, Papaioannou VE. RAG-1-deficient mice have no mature B and T lymphocytes. Cell. 1992 Mar 6;68(5):869–77. doi: 10.1016/0092-8674(92)90030-g. [DOI] [PubMed] [Google Scholar]
- 101.Shinkai Y, Rathbun G, Lam KP, Oltz EM, Stewart V, Mendelsohn M, et al. RAG-2-deficient mice lack mature lymphocytes owing to inability to initiate V(D)J rearrangement. Cell. 1992 Mar 6;68(5):855–67. doi: 10.1016/0092-8674(92)90029-c. [DOI] [PubMed] [Google Scholar]
- 102*.Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell. 2006 Jan 27;124(2):263–6. doi: 10.1016/j.cell.2006.01.007. One of the first papers to show that immune cells are important players in the metastatic cascade. [DOI] [PubMed] [Google Scholar]
- 103.Monteiro AC, Leal AC, Goncalves-Silva T, Mercadante AC, Kestelman F, Chaves SB, et al. T cells induce pre-metastatic osteolytic disease and help bone metastases establishment in a mouse model of metastatic breast cancer. PloS one. 2013;8(7):e68171. doi: 10.1371/journal.pone.0068171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kloss CC, Condomines M, Cartellieri M, Bachmann M, Sadelain M. Combinatorial antigen recognition with balanced signaling promotes selective tumor eradication by engineered T cells. Nature biotechnology. 2013 Jan;31(1):71–5. doi: 10.1038/nbt.2459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Pienta KJ, Abate-Shen C, Agus DB, Attar RM, Chung LW, Greenberg NM, et al. The current state of preclinical prostate cancer animal models. The Prostate. 2008 May 1;68(6):629–39. doi: 10.1002/pros.20726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kaighn ME, Narayan KS, Ohnuki Y, Lechner JF, Jones LW. Establishment and characterization of a human prostatic carcinoma cell line (PC-3) Investigative urology. 1979 Jul;17(1):16–23. [PubMed] [Google Scholar]
- 107.Horoszewicz JS, Leong SS, Chu TM, Wajsman ZL, Friedman M, Papsidero L, et al. The LNCaP cell line--a new model for studies on human prostatic carcinoma. Progress in clinical and biological research. 1980;37:115–32. [PubMed] [Google Scholar]
- 108.Thalmann GN, Anezinis PE, Chang SM, Zhau HE, Kim EE, Hopwood VL, et al. Androgen-independent cancer progression and bone metastasis in the LNCaP model of human prostate cancer. Cancer research. 1994 May 15;54(10):2577–81. [PubMed] [Google Scholar]
- 109.Thalmann GN, Sikes RA, Wu TT, Degeorges A, Chang SM, Ozen M, et al. LNCaP progression model of human prostate cancer: androgen-independence and osseous metastasis. The Prostate. 2000 Jul 1;44(2):91–103. doi: 10.1002/1097-0045(20000701)44:2<91::aid-pros1>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 110.Korenchuk S, Lehr JE, MC L, Lee YG, Whitney S, Vessella R, et al. VCaP, a cell-based model system of human prostate cancer. In vivo. 2001 Mar-Apr;15(2):163–8. [PubMed] [Google Scholar]
- 111.Navone NM, Olive M, Ozen M, Davis R, Troncoso P, Tu SM, et al. Establishment of two human prostate cancer cell lines derived from a single bone metastasis. Clinical cancer research : an official journal of the American Association for Cancer Research. 1997 Dec;3(12 Pt 1):2493–500. [PubMed] [Google Scholar]
- 112.Stone KR, Mickey DD, Wunderli H, Mickey GH, Paulson DF. Isolation of a human prostate carcinoma cell line (DU 145) International journal of cancer Journal international du cancer. 1978 Mar 15;21(3):274–81. doi: 10.1002/ijc.2910210305. [DOI] [PubMed] [Google Scholar]
- 113.Ito R, Takahashi T, Katano I, Ito M. Current advances in humanized mouse models. Cellular & molecular immunology. 2012 May;9(3):208–14. doi: 10.1038/cmi.2012.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Coccia MA, Brams P. High titer, prostate specific antigen-specific human IgG production by hu-PBL-SCID mice immunized with antigen-mouse IgG2a complex-pulsed autologous dendritic cells. Journal of immunology. 1998 Nov 15;161(10):5772–80. [PubMed] [Google Scholar]
- 115.Dubrovska A, Kim C, Elliott J, Shen W, Kuo TH, Koo DI, et al. A chemically induced vaccine strategy for prostate cancer. ACS chemical biology. 2011 Nov 18;6(11):1223–31. doi: 10.1021/cb200222s. [DOI] [PubMed] [Google Scholar]
- 116.Koo GC, Hasan A, O’Reilly RJ. Use of humanized severe combined immunodeficient mice for human vaccine development. Expert review of vaccines. 2009 Jan;8(1):113–20. doi: 10.1586/14760584.8.1.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117*.Lin D, Wyatt AW, Xue H, Wang Y, Dong X, Haegert A, et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer research. 2014 Feb 15;74(4):1272–83. doi: 10.1158/0008-5472.CAN-13-2921-T. Details new PDX models that coule be important in the future of drug discovery in prostate cancer. [DOI] [PubMed] [Google Scholar]
- 118.Chen X, Liu B, Li Q, Honorio S, Liu X, Liu C, et al. Dissociated primary human prostate cancer cells coinjected with the immortalized Hs5 bone marrow stromal cells generate undifferentiated tumors in NOD/SCID-gamma mice. PloS one. 2013;8(2):e56903. doi: 10.1371/journal.pone.0056903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Godebu E, Muldong M, Strasner A, Wu CN, Park SC, Woo JR, et al. PCSD1, a new patient-derived model of bone metastatic prostate cancer, is castrate-resistant in the bone-niche. Journal of translational medicine. 2014;12(1):275. doi: 10.1186/s12967-014-0275-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Raheem O, Kulidjian AA, Wu C, Jeong YB, Yamaguchi T, Smith KM, et al. A novel patient-derived intra-femoral xenograft model of bone metastatic prostate cancer that recapitulates mixed osteolytic and osteoblastic lesions. Journal of translational medicine. 2011;9:185. doi: 10.1186/1479-5876-9-185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zhao H, Thong A, Nolley R, Reese SW, Santos J, Ingels A, et al. Patient-derived tissue slice grafts accurately depict response of high-risk primary prostate cancer to androgen deprivation therapy. Journal of translational medicine. 2013;11:199. doi: 10.1186/1479-5876-11-199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122*.Russell PJ, Russell P, Rudduck C, Tse BW, Williams ED, Raghavan D. Establishing prostate cancer patient derived xenografts: Lessons learned from older studies. The Prostate. 2015 May;75(6):628–36. doi: 10.1002/pros.22946. Discusses how to make better PDXs using lessons from past models. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Rosfjord E, Lucas J, Li G, Gerber HP. Advances in patient-derived tumor xenografts: from target identification to predicting clinical response rates in oncology. Biochemical pharmacology. 2014 Sep 15;91(2):135–43. doi: 10.1016/j.bcp.2014.06.008. [DOI] [PubMed] [Google Scholar]
- 124.Corey E, Quinn JE, Buhler KR, Nelson PS, Macoska JA, True LD, et al. LuCaP 35: a new model of prostate cancer progression to androgen independence. The Prostate. 2003 Jun 1;55(4):239–46. doi: 10.1002/pros.10198. [DOI] [PubMed] [Google Scholar]
- 125.Corey E, Quinn JE, Vessella RL. A novel method of generating prostate cancer metastases from orthotopic implants. The Prostate. 2003 Jul 1;56(2):110–4. doi: 10.1002/pros.10235. [DOI] [PubMed] [Google Scholar]
- 126.Ellis WJ, Vessella RL, Buhler KR, Bladou F, True LD, Bigler SA, et al. Characterization of a novel androgen-sensitive, prostate-specific antigen-producing prostatic carcinoma xenograft: LuCaP 23. Clinical cancer research : an official journal of the American Association for Cancer Research. 1996 Jun;2(6):1039–48. [PubMed] [Google Scholar]
- 127.Young SR, Saar M, Santos J, Nguyen HM, Vessella RL, Peehl DM. Establishment and serial passage of cell cultures derived from LuCaP xenografts. The Prostate. 2013 Sep;73(12):1251–62. doi: 10.1002/pros.22610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.McCulloch DR, Opeskin K, Thompson EW, Williams ED. BM18: A novel androgen-dependent human prostate cancer xenograft model derived from a bone metastasis. The Prostate. 2005 Sep 15;65(1):35–43. doi: 10.1002/pros.20255. [DOI] [PubMed] [Google Scholar]
- 129.Aparicio A, Tzelepi V, Araujo JC, Guo CC, Liang S, Troncoso P, et al. Neuroendocrine prostate cancer xenografts with large-cell and small-cell features derived from a single patient’s tumor: morphological, immunohistochemical, and gene expression profiles. The Prostate. 2011 Jun 1;71(8):846–56. doi: 10.1002/pros.21301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Fong EL, Martinez M, Yang J, Mikos AG, Navone NM, Harrington DA, et al. Hydrogel-based 3D model of patient-derived prostate xenograft tumors suitable for drug screening. Molecular pharmaceutics. 2014 Jul 7;11(7):2040–50. doi: 10.1021/mp500085p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lin D, Xue H, Wang Y, Wu R, Watahiki A, Dong X, et al. Next generation patient-derived prostate cancer xenograft models. Asian journal of andrology. 2014 May-Jun;16(3):407–12. doi: 10.4103/1008-682X.125394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wetterauer C, Vlajnic T, Schuler J, Gsponer JR, Thalmann GN, Cecchini M, et al. Early development of human lymphomas in a prostate cancer xenograft program using triple knock-out Immunocompromised mice. The Prostate. 2015 May;75(6):585–92. doi: 10.1002/pros.22939. [DOI] [PubMed] [Google Scholar]
- 133.Ellis L, Lehet K, Ku S, Azabdaftari G, Pili R. Generation of a syngeneic orthotopic transplant model of prostate cancer metastasis. Oncoscience. 2014 Oct 15;1(10):609–13. doi: 10.18632/oncoscience.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Liao CP, Liang M, Cohen MB, Flesken-Nikitin A, Jeong JH, Nikitin AY, et al. Mouse prostate cancer cell lines established from primary and postcastration recurrent tumors. Hormones & cancer. 2010 Feb;1(1):44–54. doi: 10.1007/s12672-009-0005-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Watson PA, Ellwood-Yen K, King JC, Wongvipat J, Lebeau MM, Sawyers CL. Context-dependent hormone-refractory progression revealed through characterization of a novel murine prostate cancer cell line. Cancer research. 2005 Dec 15;65(24):11565–71. doi: 10.1158/0008-5472.CAN-05-3441. [DOI] [PubMed] [Google Scholar]
- 136.Haga K, Tomioka A, Liao CP, Kimura T, Matsumoto H, Ohno I, et al. PTEN knockout prostate cancer as a model for experimental immunotherapy. The Journal of urology. 2009 Jan;181(1):354–62. doi: 10.1016/j.juro.2008.08.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Liu Z, Eltoum IE, Guo B, Beck BH, Cloud GA, Lopez RD. Protective immunosurveillance and therapeutic antitumor activity of gammadelta T cells demonstrated in a mouse model of prostate cancer. Journal of immunology. 2008 May 1;180(9):6044–53. doi: 10.4049/jimmunol.180.9.6044. [DOI] [PubMed] [Google Scholar]
- 138.Saika T, Kusaka N, Mouraviev V, Satoh T, Kumon H, Timme TL, et al. Therapeutic effects of adoptive splenocyte transfer following in situ AdIL-12 gene therapy in a mouse prostate cancer model. Cancer gene therapy. 2006 Jan 1;13(1):91–8. doi: 10.1038/sj.cgt.7700872. [DOI] [PubMed] [Google Scholar]
- 139.Tse BW, Russell PJ, Lochner M, Forster I, Power CA. IL-18 inhibits growth of murine orthotopic prostate carcinomas via both adaptive and innate immune mechanisms. PloS one. 2011;6(9):e24241. doi: 10.1371/journal.pone.0024241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Tseng-Rogenski SS, Arredouani MS, Neeley YC, Lu B, Chinnaiyan AM, Sanda MG. Fas-mediated T cell deletion potentiates tumor antigen-specific tolerance in a mouse model of prostate cancer. Cancer immunology, immunotherapy : CII. 2008 Sep;57(9):1357–65. doi: 10.1007/s00262-008-0471-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Havens AM, Pedersen EA, Shiozawa Y, Ying C, Jung Y, Sun Y, et al. An in vivo mouse model for human prostate cancer metastasis. Neoplasia. 2008 Apr;10(4):371–80. doi: 10.1593/neo.08154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Hutchinson L, Kirk R. High drug attrition rates--where are we going wrong? Nature reviews Clinical oncology. 2011 Apr;8(4):189–90. doi: 10.1038/nrclinonc.2011.34. [DOI] [PubMed] [Google Scholar]
- 143.An Z, Wang X, Geller J, Moossa AR, Hoffman RM. Surgical orthotopic implantation allows high lung and lymph node metastatic expression of human prostate carcinoma cell line PC-3 in nude mice. The Prostate. 1998 Feb 15;34(3):169–74. doi: 10.1002/(sici)1097-0045(19980215)34:3<169::aid-pros3>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
- 144.Glinskii AB, Smith BA, Jiang P, Li XM, Yang M, Hoffman RM, et al. Viable circulating metastatic cells produced in orthotopic but not ectopic prostate cancer models. Cancer research. 2003 Jul 15;63(14):4239–43. [PubMed] [Google Scholar]
- 145.Kolostova K, Pinterova D, Hoffman RM, Bobek V. Circulating human prostate cancer cells from an orthotopic mouse model rapidly captured by immunomagnetic beads and imaged by GFP expression. Anticancer research. 2011 May;31(5):1535–9. [PubMed] [Google Scholar]
- 146.Ongkeko WM, Burton D, Kiang A, Abhold E, Kuo SZ, Rahimy E, et al. Parathyroid hormone related-protein promotes epithelial-to-mesenchymal transition in prostate cancer. PloS one. 2014;9(1):e85803. doi: 10.1371/journal.pone.0085803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Wang X, An Z, Geller J, Hoffman RM. High-malignancy orthotopic nude mouse model of human prostate cancer LNCaP. The Prostate. 1999 May 15;39(3):182–6. doi: 10.1002/(sici)1097-0045(19990515)39:3<182::aid-pros6>3.0.co;2-b. [DOI] [PubMed] [Google Scholar]
- 148*.de Jong M, Maina T. Of mice and humans: are they the same?--Implications in cancer translational research. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2010 Apr;51(4):501–4. doi: 10.2967/jnumed.109.065706. Important questions regarding the use of mice in human disease research. [DOI] [PubMed] [Google Scholar]
- 149.Stahl WR. Scaling of respiratory variables in mammals. Journal of applied physiology. 1967 Mar;22(3):453–60. doi: 10.1152/jappl.1967.22.3.453. [DOI] [PubMed] [Google Scholar]
- 150.Missaghian E, Kempna P, Dick B, Hirsch A, Alikhani-Koupaei R, Jegou B, et al. Role of DNA methylation in the tissue-specific expression of the CYP17A1 gene for steroidogenesis in rodents. The Journal of endocrinology. 2009 Jul;202(1):99–109. doi: 10.1677/JOE-08-0353. [DOI] [PubMed] [Google Scholar]
- 151.Cai C, Chen S, Ng P, Bubley GJ, Nelson PS, Mostaghel EA, et al. Intratumoral de novo steroid synthesis activates androgen receptor in castration-resistant prostate cancer and is upregulated by treatment with CYP17A1 inhibitors. Cancer research. 2011 Oct 15;71(20):6503–13. doi: 10.1158/0008-5472.CAN-11-0532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Mostaghel EA, Marck BT, Plymate SR, Vessella RL, Balk S, Matsumoto AM, et al. Resistance to CYP17A1 inhibition with abiraterone in castration-resistant prostate cancer: induction of steroidogenesis and androgen receptor splice variants. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011 Sep 15;17(18):5913–25. doi: 10.1158/1078-0432.CCR-11-0728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.van der Worp HB, Howells DW, Sena ES, Porritt MJ, Rewell S, O’Collins V, et al. Can animal models of disease reliably inform human studies? PLoS medicine. 2010 Mar;7(3):e1000245. doi: 10.1371/journal.pmed.1000245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Hackam DG, Redelmeier DA. Translation of research evidence from animals to humans. Jama. 2006 Oct 11;296(14):1731–2. doi: 10.1001/jama.296.14.1731. [DOI] [PubMed] [Google Scholar]
- 155.Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ, et al. Guidelines for the welfare and use of animals in cancer research. British journal of cancer. 2010 May 25;102(11):1555–77. doi: 10.1038/sj.bjc.6605642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Sena ES, van der Worp HB, Bath PM, Howells DW, Macleod MR. Publication bias in reports of animal stroke studies leads to major overstatement of efficacy. PLoS biology. 2010 Mar;8(3):e1000344. doi: 10.1371/journal.pbio.1000344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157**.Perrin S. Preclinical research: Make mouse studies work. Nature. 2014 Mar 27;507(7493):423–5. doi: 10.1038/507423a. Critical review of the use of mouse models, where we have gone wrong, and the need to make mouse models work better to predict drugs’ effectiveness in human. [DOI] [PubMed] [Google Scholar]
- 158.Begley CG, Ellis LM. Drug development: Raise standards for preclinical cancer research. Nature. 2012 Mar 29;483(7391):531–3. doi: 10.1038/483531a. [DOI] [PubMed] [Google Scholar]
- 159.Conway JR, Carragher NO, Timpson P. Developments in preclinical cancer imaging: innovating the discovery of therapeutics. Nature reviews Cancer. 2014 May;14(5):314–28. doi: 10.1038/nrc3724. [DOI] [PubMed] [Google Scholar]
- 160.Yang M, Jiang P, Sun FX, Hasegawa S, Baranov E, Chishima T, et al. A fluorescent orthotopic bone metastasis model of human prostate cancer. Cancer research. 1999 Feb 15;59(4):781–6. [PubMed] [Google Scholar]
- 161.Yang M, Jiang P, Yamamoto N, Li L, Geller J, Moossa AR, et al. Real-time whole-body imaging of an orthotopic metastatic prostate cancer model expressing red fluorescent protein. The Prostate. 2005 Mar 1;62(4):374–9. doi: 10.1002/pros.20125. [DOI] [PubMed] [Google Scholar]
- 162.Abate-Shen C, Shen MM. Molecular genetics of prostate cancer. Genes & development. 2000 Oct 1;14(19):2410–34. doi: 10.1101/gad.819500. [DOI] [PubMed] [Google Scholar]
- 163.McNeal JE. Origin and development of carcinoma in the prostate. Cancer. 1969 Jan;23(1):24–34. doi: 10.1002/1097-0142(196901)23:1<24::aid-cncr2820230103>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
